Suppressing ROS generation by apocynin inhibited cyclic stretch-induced inflammatory reaction in HPDLCs via a caspase-1 dependent pathway

https://doi.org/10.1016/j.intimp.2020.107129Get rights and content

Highlights

  • This study inspected the role of ROS in the cyclic stretch-induced inflammatory reaction in HPDLCs.

  • The results indicated that cyclic stretch induced inflammatory reaction in HPDLCs via the regulation of ROS/NLRP3/caspase-1 signaling pathway.

  • It was found in the present study that both the stretch-induced inflammatory reaction and the expression of caspase-1 and NLRP3 in HPDLCs could be inhibited by antioxidant apocynin.

  • This study put forward the possibility of antioxidant apocynin as one treatment for stretch-induced inflammatory reaction in HPDLCs.

Abstract

It has been reported that cyclic stretch could induce inflammatory reaction in human periodontal ligament cells (HPDLCs). Though reactive oxygen species (ROS) has been reported to be involved in pathogen-induced periodontal inflammatory reaction, its role in the force-related periodontal diseases has not been well clarified. This study inspected the role of ROS in the cyclic stretch-induced inflammatory reaction in HPDLCs and studied the inhibitory effect of antioxidant apocynin on this inflammatory reaction. Results confirmed that cyclic stretch induced inflammatory reaction and production of ROS in HPDLCs. This inflammatory reaction was inhibited by apocynin through blocking the production of ROS. The cyclic stretch also induced the expression of caspase-1 and NLRP3 inflammasome, which could also be inhibited by apocynin. Moreover, the cyclic stretch-induced inflammatory reaction was inhibited by caspase-1 inhibitor. Collectively, it is the first time that increased intracellular ROS was proved to play as an intermediate signal in the cyclic stretch-induced inflammatory reaction in HPDLCs, via a caspase-1-dependent pathway. The inhibitory effect of apocynin on the cyclic stretch-induced inflammatory reaction in HPDLCs shows the potential of antioxidants in the treatment of force-related periodontal inflammatory diseases.

Introduction

Periodontal ligament (PDL) is the primary target of mechanical forces aroused during occlusion, mastication and orthodontic treatment [1], [2], [3]. Human periodontal ligament cells (HPDLCs) are the sensors of such mechanical stimuli and respond to them through activated metabolism, proliferation, osteoblastic differentiation and inflammation, thus play an important role in biological processes of the maintenance of periodontal tissue homeostasis, repair, and remodelling [1], [2]. It has been reported that HPDLCs also participated in some pathological process such as mechanical force-induced periodontitis, which was aroused by abnormal mechanical stimulations from occlusal overloading or improper orthodontic treatment [4], [5], [6], [7], [8], [9].

Our recent study showed that cyclic stretch activated Nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome and induced inflammatory reaction including the secretion of pro-inflammation cytokines and pyroptosis in HPDLCs [10], and confirmed the role of NLRP3 inflammasome in the mechanical force-induced periodontal inflammatory reaction. NLRP3 inflammasome is a high-molecular-weight oligomer formed by oligomerization of NLRP3, ASC and caspase-1 [11], and has been reported to play a critical role in the development of periodontitis [12]. In the oligomer, pro-caspase-1 is converted to its active form, caspase-1, which will subsequently cleave its substrates such as pro-interleukin-1β (pro-IL-1β) and pro-interleukin-18 (pro-IL-18) to their bioactive forms (IL-1β and IL-18) [13].

Several cellular events, such as mitochondrial dysfunction, reactive oxygen species (ROS) production, potassium efflux, and cell swelling, have been proposed as upstream signals for the activation of NLRP3 inflammasome [14], [15], [16]. It has been reported that lipopolysaccharide (LPS) and advanced products of glycation end activated NLRP3 inflammasome and induced inflammatory reaction in HPDLCs through production of ROS [11], [17]. These findings confirmed the involvement of ROS in the pathogen-related activation of inflammasome and the pathogen-induced periodontal inflammatory reaction. Whether ROS are involved in force-related activation of inflammasomes and force-induced periodontal inflammatory reaction still remains unclear. Interestingly, a recent study confirmed the activation of NLRP3 inflammasome in mechanical stretch-induced lung injury via ROS production [18]. Furthermore, another recent study showed that ROS level in capillary blood increased after short-term (24 h) orthodontic force loading in patients treated with fixed orthodontic appliances [19]. Therefore, it’s reasonable to speculate that ROS might also play a role in force-related activation of inflammasomes and force-induced periodontal inflammatory reaction.

ROS are considered to be a double-edged sword in periodontal diseases [20]. At low concentrations, ROS stimulate the proliferation and differentiation of HPDLCs, while at higher concentrations, they may have cytotoxic effects on periodontal tissues [21], [22], [23], [24], [25]. Oxidative stress or ROS accumulation is a significant process in the pathogenesis of several oral diseases [26], [27].

Previous studies have found that ROS are produced in the mitochondrial respiratory chain during cellular respiration and oxidative reactions which are catalyzed by NADPH oxidase, xanthine oxidase or Lamino-acid-oxidase [28]. The key producer of ROS in many cells is NADPH oxidase, comprising membrane and cytosolic components, which actively communicates during the host responses to a wide variety of stimuli [29]. NADPH Oxidase is a multi-subunit complex composed of six subunits [29]. Apocynin (4′-hydroxy-3′-methoxyacetophenone or acetovanillone) can block the activity of NADPH oxidase by preventing the phosphorylation of the p47phox subunit of NADPH Oxidase [30]. Many studies have shown the anti-inflammatory and antioxidation effects of apocynin in various human diseases, including LPS induced periodontal diseases [11]. However, the role of ROS in cyclic stretch-induced inflammatory reaction in HPDLCs still remained to be elucidated.

Taken together, we hypothesize that cyclic stretch induces inflammatory reaction in HPDLCs via a ROS-related signaling pathway and apocynin as ROS inhibitor can help suppress this cyclic stretch-induced inflammatory reaction. The findings of this study may help us better understand the mechanism of the mechanical force-related periodontal inflammation and provide evidences to support antioxidants as a hopeful treatment for these kinds of periodontal diseases.

Section snippets

Cell cultures and cyclic stretch loading

HPDLCs were cultured following the method reported by us previously, which has been described by Zhuang et al. [31] in detail. In brief, pieces of PDL tissues were harvested from the middle of the root surface of extracted premolars of teenagers (11–16 years old) for orthodontic reason with the informed consents from their parents. Collected PDL tissues were attached to a cell culture dish and incubated with high-glucose Dulbecco’s modification of Eagle’s medium (DMEM, HyClone, Logan, UT, USA)

Cyclic stretch induced the generation of ROS in HPDLCs

Result of cellular ROS detection by Flow Cytometry showed that the expression of ROS in HPDLCs increased in response to 3 h and 6 h cyclic stretch (P < 0.01 VS control for stretched-3 h group, P < 0.001 VS control for stretched-6 h group) and it went down after 12 h and 24 h cyclic stretching (P < 0.001 VS stretched-6 h group) (Fig. 1).

Cyclic stretch induced pyroptosis in HPDLCs

Flow cytometric analysis demonstrated that the pyroptotic rate of HPDLCs in response to 6 h cyclic stretch increased significantly, compared with non-stretched

Discussion

The PDL functions as a cushion to withstand mechanical forces applied to teeth. PDLCs are stimulated by forces applied during mastication, occlusal contact, and orthodontic treatment and produce local factors that participate not only in the maintenance and remodeling of the ligament itself, but also in the metabolism of adjacent alveolar bone [35]. Physiological mechanical forces help to maintain the balance in periodontal tissue, while abnormal mechanical forces such as traumatic occlusion

Conclusions

The present study showed for the first time that the cyclic stretch induced inflammatory reaction in HPDLCs through stimulating the production of ROS, probably via a caspase-1-dependent mechanism. Apocynin could be a promising option for protecting periodontal tissue against force-related inflammation. Nevertheless, the exact regulating mechanisms in this force-related ROS/NLRP3/caspase-1 signaling pathway need to be further elucidated.

Funding

This work was supported by National Natural Science Foundation of China (grant numbers 31470903, 31270991, 30900282), Shanghai Municipal Health Commission (grant number 201940009), Shanghai Summit & Plateau Disciplines, Shanghai Pujiang Program (grant number 13PJD021), and Science and Technology Commission of Shanghai (grant number 10QA1404200, 08411961500, 07ZR14070).

Compliance with Ethical Standards

The experimental protocol was reviewed and approved by the Ethics Committee of Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine. The relevant judgment’s reference number is [2017]96.

CRediT authorship contribution statement

Yingying Wang: Conceptualization, Data curation, Investigation, Methodology, Software, Writing - original draft, Writing - review & editing. Yingshuang Song: Data curation, Visualization. Qi Zhong: Data curation, Software. Yaqin Wu: Conceptualization, Methodology. Jiabao Zhuang: Methodology, Supervision. Fang Qu: Conceptualization, Supervision. Chun Xu: Conceptualization, Funding acquisition, Resources, Writing - review & editing.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (61)

  • R. Xin et al.

    Apocynin inhibited NLRP3/XIAP signalling to alleviate renal fibrotic injury in rat diabetic nephropathy

    Biomed. Pharmacotherapie

    (2018)
  • X. Liu et al.

    Enhanced efficacy of baicalin-loaded TPGS polymeric micelles against periodontitis

    Mater. Sci. Eng. C, Mater. Biol. Appl.

    (2019)
  • P. Lekic et al.

    Periodontal ligament cell population: the central role of fibroblasts in creating a unique tissue

    Anatomical Rec.

    (1996)
  • G. Isola et al.

    Mechanobiology of the tooth movement during the orthodontic treatment: a literature review

    Minervastomatologica

    (2016)
  • C. Jacobs et al.

    Mechanical loading increases pro-inflammatory effects of nitrogen-containing bisphosphonate in human periodontal fibroblasts

    Clin. Oral Invest.

    (2018)
  • N. Arceo et al.

    Human periodontal cells initiate mineral-like nodules in vitro

    J. Periodontol.

    (1991)
  • D.L. Carnes et al.

    Cells with osteoblastic phenotypes can be explanted from human gingiva and periodontal ligament

    J. Periodontol.

    (1997)
  • A. Papadopoulou et al.

    Effect of hyperglycaemic conditions on the response of human periodontal ligament fibroblasts to mechanical stretching

    Eur. J. Orthod.

    (2019)
  • M. Kaku et al.

    Investigation of periodontal ligament reaction upon excessive occlusal load–osteopontin induction among periodontal ligament cells

    J. Periodontal Res.

    (2005)
  • D. Zhao et al.

    Activation of NLRP1 and NLRP3 inflammasomes contributed to cyclic stretch-induced pyroptosis and release of IL-1beta in human periodontal ligament cells

    Oncotarget

    (2016)
  • K. Shibata

    Historical aspects of studies on roles of the inflammasome in the pathogenesis of periodontal diseases

    Mol. Oral Microbiol.

    (2018)
  • Z. Ma et al.

    Editorial: NLRP3: immune activator or modulator?

    J. Leukoc. Biol.

    (2016)
  • Lamkanfi, M. D., V.M. Mechanisms and functions of inflammasomes. Cell 2014, 157,...
  • P. Broz et al.

    Inflammasomes: mechanism of assembly, regulation and signalling

    Nat. Rev. Immunol.

    (2016)
  • K.H. Lee et al.

    The molecular links between cell death and inflammasome

    Cells

    (2019)
  • Y.M. Mei et al.

    AGEs induces apoptosis and autophagy via reactive oxygen species in human periodontal ligament cells

    J. Cell. Biochem.

    (2019)
  • Wu J, Y. Z., Schwartz DE, et al. Activation of NLRP3 inflammasome in alveolar macrophages contributes to mechanical...
  • V. Kovac et al.

    Systemic level of oxidative stress during orthodontic treatment with fixed appliances

    BioMed Res. Int.

    (2019)
  • L. Nibali et al.

    Periodontitis and redox status: a review

    Curr. Pharm. Des.

    (2013)
  • I.L. Chapple et al.

    The role of reactive oxygen and antioxidant species in periodontal tissue destruction

    Periodontology

    (2000)
  • Cited by (0)

    1

    Yingying Wang and Yingshuang Song should be considered joint first author.

    View full text